Skeletal responses to romosozumab after 12 months of denosumab

Michael R McClung, Michael A Bolognese, Jacques P Brown, Jean-Yves Reginster, Bente L Langdahl, Yifei Shi, Jen Timoshanko, Cesar Libanati, Arkadi Chines, Mary K Oates, Michael R McClung, Michael A Bolognese, Jacques P Brown, Jean-Yves Reginster, Bente L Langdahl, Yifei Shi, Jen Timoshanko, Cesar Libanati, Arkadi Chines, Mary K Oates

Abstract

Romosozumab, a monoclonal anti-sclerostin antibody that has the dual effect of increasing bone formation and decreasing bone resorption, reduces fracture risk within 12 months. In a post hoc, exploratory analysis, we evaluated the effects of romosozumab after 12 months of denosumab in postmenopausal women with low bone mass who had not received previous osteoporosis therapy. This phase 2 trial (NCT00896532) enrolled postmenopausal women with a lumbar spine, total hip, or femoral neck T-score ≤ -2.0 and ≥ -3.5. Individuals were randomized to placebo or various romosozumab dosing regimens from baseline to month 24, were re-randomized to 12 months of denosumab or placebo (months 24-36), and then all received romosozumab 210 mg monthly for 12 months (months 36-48). Results for the overall population have been previously published. Here, we present results for changes in bone mineral density (BMD) and levels of procollagen type I N-terminal propeptide (P1NP) and β-isomer of the C-terminal telopeptide of type I collagen (β-CTX) from a subset of women who were randomized to placebo for 24 months, were re-randomized to receive denosumab (n = 16) or placebo (n = 12) for 12 months, and then received romosozumab for 12 months. In women who were randomized to placebo followed by denosumab, romosozumab treatment for 12 months maintained BMD gained during denosumab treatment at the total hip (mean change from end of denosumab treatment of 0.9%) and further increased BMD gains at the lumbar spine (mean change from end of denosumab treatment of 5.3%). Upon transition to romosozumab (months 36-48), P1NP and β-CTX levels gradually returned to baseline from their reduced values during denosumab administration. Transitioning to romosozumab after 12 months of denosumab appears to improve lumbar spine BMD and maintain total hip BMD while possibly preventing the rapid increase in levels of bone turnover markers above baseline expected upon denosumab discontinuation. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

Keywords: ANABOLIC; ANTIRESORPTIVE; DENOSUMAB; ROMOSOZUMAB; TREATMENT SEQUENCE.

© 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

Figures

Fig. 1
Fig. 1
Phase 2 study design. (A) Women were randomized 1:1:1:1:1:1:1:1 to the first 24 months of treatment. Administration of placebo and the various romosozumab doses was blinded; alendronate and teriparatide were administered open‐label. At month 24, women were rerandomized (1:1) within treatment group to placebo or denosumab (60 mg s.c. Q6M) for 12 months, followed by a 12‐month second course of romosozumab 210 mg s.c. QM. (B) A subset of women who were randomized to receive placebo for 24 months (n = 52), rerandomized to receive denosumab (n = 16) or placebo (n = 12) for 12 months, and then received romosozumab for 12 months and whose results are presented in this report. aIndividuals transitioned to romosozumab 140 mg QM at month 12, were randomized in the denosumab extension period, completed the study at month 36 and are not included in the present analysis. bIndividuals completed the study at month 12 and are not included in the present analysis. cOf the 52 women initially randomized to placebo from months 0 to 24, 18 were rerandomized to receive denosumab and 18 to receive placebo; the remaining 16 discontinued the study. Abbreviations: Q3M, every 3 months; Q6M, every 6 months; QD, daily; QM, monthly; QW, weekly; s.c., subcutaneously.
Fig. 2
Fig. 2
Percentage change from baseline in lumbar spine and total hip BMD through month 48 for (A,C) placebo‐to‐placebo‐to‐romosozumab and (B,D) placebo‐to‐denosumab‐to‐romosozumab. Data reported are for a subset of women who were randomized to receive placebo for 24 months (n = 52), rerandomized to receive denosumab or placebo for 12 months, and then received romosozumab for 12 months. n = number of women enrolled from month 36 to month 48. Abbreviations: BMD, bone mineral density; Q6M, every 6 months; QM, monthly.
Fig. 3
Fig. 3
Percentage changes from baseline in serum P1NP and β‐CTX through month 48 for (A,C) placebo‐to‐placebo‐to‐romosozumab and (B,D) placebo‐to‐denosumab‐to‐romosozumab. Data reported are for a subset of women who were randomized to receive placebo for 24 months (n = 52), rerandomized to receive denosumab or placebo for 12 months, and then received romosozumab for 12 months. n = number of women enrolled from month 36 to month 48. Abbreviations: β‐CTX, β‐isomer of the C‐terminal telopeptide of type I collagen; P1NP, procollagen type I N‐terminal propeptide; Q1, first quartile; Q3, third quartile; Q6M, every 6 months; QM, monthly.

References

    1. Compston JE, McClung MR, Leslie WD. Osteoporosis. Lancet. 2019;393:364‐376.
    1. Dufresne TE, Chmielewski PA, Manhart MD, Johnson TD, Borah B. Risedronate preserves bone architecture in early postmenopausal women in 1 year as measured by three‐dimensional microcomputed tomography. Calcif Tissue Int. 2003;73:423‐432.
    1. Saag KG, Petersen J, Brandi ML, et al. Romosozumab or alendronate for fracture prevention in women with osteoporosis. N Engl J Med. 2017;377:1417‐1427.
    1. Kendler DL, Marin F, Zerbini CAF, et al. Effects of teriparatide and risedronate on new fractures in post‐menopausal women with severe osteoporosis (VERO): a multicentre, double‐blind, double‐dummy, randomised controlled trial. Lancet. 2018;391:230‐240.
    1. Chavassieux P, Chapurlat R, Portero‐Muzy N, et al. Bone‐forming and antiresorptive effects of romosozumab in postmenopausal women with osteoporosis: bone histomorphometry and microcomputed tomography analysis after 2 and 12 months of treatment. J Bone Miner Res. 2019;34:1597‐1608.
    1. Jiang Y, Zhao JJ, Mitlak BH, et al. Recombinant human parathyroid hormone (1‐34) [teriparatide] improves both cortical and cancellous bone structure. J Bone Miner Res. 2003;18:1932‐1941.
    1. Miller PD, Hattersley G, Riis BJ, et al. Effect of abaloparatide vs placebo on new vertebral fractures in postmenopausal women with osteoporosis: a randomized clinical trial. JAMA. 2016;316:722‐733.
    1. Shoback D, Rosen CJ, Black DM, et al. Pharmacological management of osteoporosis in postmenopausal women: an Endocrine Society guideline update. J Clin Endocrinol Metab. 2020;105:587‐594.
    1. Camacho PM, Petak SM, Binkley N, et al. American Association of Clinical Endocrinologists/American College of Endocrinology clinical practice guidelines for the diagnosis and treatment of postmenopausal osteoporosis ‐ 2020 update. Endocr Pract. 2020;26:1‐46.
    1. Kanis JA, Harvey NC, McCloskey E, et al. Algorithm for the management of patients at low, high and very high risk of osteoporotic fractures. Osteoporos Int. 2020;31:1‐12.
    1. Ominsky MS, Niu Q‐T, Li C, Li X, Ke HZ. Tissue‐level mechanisms responsible for the increase in bone formation and bone volume by sclerostin antibody. J Bone Miner Res. 2014;29:1424‐1430.
    1. McClung MR. Romosozumab for the treatment of osteoporosis. Osteoporos Sarcopenia. 2018;4:11‐15.
    1. Cosman F, Crittenden DB, Adachi JD, et al. Romosozumab treatment in postmenopausal women with osteoporosis. N Engl J Med. 2016;375:1532‐1543.
    1. McClung MR, Grauer A, Boonen S, et al. Romosozumab in postmenopausal women with low bone mineral density. N Engl J Med. 2014;370:412‐420.
    1. Cosman F, Crittenden DB, Ferrari S, et al. FRAME study: the foundation effect of building bone with 1 year of romosozumab leads to continued lower fracture risk after transition to denosumab. J Bone Miner Res. 2018;33:1219‐1226.
    1. Langdahl BL, Libanati C, Crittenden DB, et al. Romosozumab (sclerostin monoclonal antibody) versus teriparatide in postmenopausal women with osteoporosis transitioning from oral bisphosphonate therapy: a randomised, open‐label, phase 3 trial. Lancet. 2017;390:1585‐1594.
    1. Kendler DL, Bone HG, Massari F, et al. Bone mineral density gains with a second 12‐month course of romosozumab therapy following placebo or denosumab. Osteoporos Int. 2019;30:2437‐2448.
    1. McClung MR, Brown JP, Diez‐Perez A, et al. Effects of 24 months of treatment with romosozumab followed by 12 months of Denosumab or placebo in postmenopausal women with low Bone mineral density: a randomized, double‐blind, phase 2, parallel group study. J Bone Miner Res. 2018;33:1397‐1406.
    1. McClung MR, Bolognese MA, Brown JP, et al. A single dose of zoledronate preserves bone mineral density for up to 2 years after a second course of romosozumab. Osteoporos Int. 2020;31:2231‐2241.
    1. Miller PD, Bolognese MA, Lewiecki EM, et al. Effect of denosumab on bone density and turnover in postmenopausal women with low bone mass after long‐term continued, discontinued, and restarting of therapy: a randomized blinded phase 2 clinical trial. Bone. 2008;43:222‐229.
    1. Bone HG, Bolognese MA, Yuen CK, et al. Effects of denosumab treatment and discontinuation on bone mineral density and bone turnover markers in postmenopausal women with low bone mass. J Clin Endocrinol Metab. 2011;96:972‐980.
    1. Leder BZ, Tsai JN, Uihlein AV, et al. Denosumab and teriparatide transitions in postmenopausal osteoporosis (the DATA‐Switch study): extension of a randomised controlled trial. Lancet. 2015;386:1147‐1155.
    1. Cosman F, Lewiecki EM, Ebeling PR, et al. T‐score as an indicator of fracture risk during treatment with romosozumab or alendronate in the ARCH trial. J Bone Miner Res. 2020;35:1333‐1342.
    1. Ferrari S, Libanati C, Lin CJF, et al. Relationship between bone mineral density T‐score and nonvertebral fracture risk over 10 years of denosumab treatment. J Bone Miner Res. 2019;34:1033‐1040.
    1. Bouxsein ML, Eastell R, Lui L‐Y, et al. Change in bone density and reduction in fracture risk: a meta‐regression of published trials. J Bone Miner Res. 2019;34:632‐642.
    1. Cosman F. Anabolic therapy and optimal treatment sequences for patients with osteoporosis at high risk for fracture. Endocr Pract. 2020;26:777‐786.
    1. Chen P, Satterwhite JH, Licata AA, et al. Early changes in biochemical markers of bone formation predict BMD response to teriparatide in postmenopausal women with osteoporosis. J Bone Miner Res. 2005;20:962‐970.
    1. Mawatari T, Ikemura S, Matsui G, et al. Assessment of baseline bone turnover marker levels and response to risedronate treatment: data from a Japanese phase III trial. Bone Rep. 2020;12:100275.
    1. Bauer DC, Garnero P, Hochberg MC, et al. Pretreatment levels of bone turnover and the antifracture efficacy of alendronate: the fracture intervention trial. J Bone Miner Res. 2006;21:292‐299.
    1. McClung MR, Lewiecki EM, Cohen SB, et al. Denosumab in postmenopausal women with low bone mineral density. N Engl J Med. 2006;354:821‐831.
    1. Eastell R, Christiansen C, Grauer A, et al. Effects of denosumab on bone turnover markers in postmenopausal osteoporosis. J Bone Miner Res. 2011;26:530‐537.
    1. Ebina K, Hirao M, Tsuboi H, et al. Effects of prior osteoporosis treatment on early treatment response of romosozumab in patients with postmenopausal osteoporosis. Bone. 2020;140:115574.

Source: PubMed

3
Subskrybuj